Bevacizumab Immune Complexes Induce Thrombocytopenia and Thrombosis in Mice Transgenic for Human IgG Receptor FcYIIa

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 114-114
Author(s):  
Ali Amirkhosravi ◽  
Todd V Meyer ◽  
Liza Robles-Carillo ◽  
Florian Langer ◽  
Theresa Robson ◽  
...  

Abstract Background: The anti-VEGF drug, bevacizumab (Bev), has been associated with arterial thromboembolism in colorectal cancer patients. However, the mechanism of this remains poorly understood, and preclinical testing in mice failed to predict thrombosis. Prevailing opinion on the molecular mechanism behind Bev-associated bleeding and thrombosis is that tissue factor driven coagulation, secondary to vascular endothelial cell dysfunction, may cause thrombosis due to VEGF suppression by Bev. Bev forms immune complexes (IC) with VEGF (vascular endothelial growth factor), a heparin-binding protein. In our previous in vitro studies we showed that, in the presence of heparin, Bev+VEGF immune complexes activate platelets via the IgG receptor FcγRIIa —a mechanism similar to that observed with antibodies from patients with heparin-induced thrombocytopenia (HIT). Objectives: First, we investigated whether Bev-associated thrombosis might be replicated in mice. Because mouse platelets do not carry FcγRIIa, we used mice transgenic for this human IgG receptor (hFcR mice) in order to enable the signaling pathway identified above. Second, using human platelets in vitro, we studied the functional roles of heparin and platelet surface localization of IC in Bev-induced FcγRIIa activation. Methods: Bev+VEGF IC were preformed using VEGF165 or VEGF121 (similar to VEGF165 but lacking the heparin-binding domain). Platelet dense granule release and aggregation were measured by the serotonin release assay (SRA) and Chrono- Log aggregometers, respectively. Platelet surface localization was assessed by flow cytometry (50,000 events/test condition) and fluorescence microscopy using Alexa488- labeled Bev (Bev488). For in vivo studies, Bev+VEGF+Heparin IC (60–500 nM) or control reagents were injected intravenously into wild-type (WT) or hFcR mice. Platelet counts were measured 10–60 minutes following IC injection after obtaining blood (0.45 ml) by cardiac puncture. Immediately afterward, lungs were processed for hematoxylin and eosin staining and analyzed microscopically for evidence of thrombosis. Results: IC consisting of Bev+VEGF165+Heparin (0.2U/ml) caused thrombotic thrombocytopenia in hFcR but not WT mice, showing a requirement for FcγRIIa. Injection of Bev+VEGF121+Heparin (0.2U/ml) into hFcR mice did not cause thrombocytopenia, suggesting a requirement for the VEGF165 heparin binding domain. Bev+VEGF165 was without effect in the absence of heparin or in the presence of excess (200 U/ml) heparin demonstrating that a limited range of heparin concentrations enable Bev-induced thrombocytopenia and thrombosis. This mechanism is similar to that observed in HIT and our in vivo results were consistent with SRA and aggregation in vitro studies. By flow cytometry, maximal Fab-dependent Bev488 platelet surface binding occured only with VEGF165+0.2U/ml heparin. Saturating IV.3 (anti-FcγRIIa antibody) concentrations, present in all samples, excluded Bev-Fc binding to FcγRIIa. Furthermore, binding of Bev488+VEGF121+0.2 U/ml heparin was not detected, suggesting the VEGF heparin binding domain is required for heparin-enhanced surface binding. Conclusions: In the presence of heparin, Bev can induce platelet aggregation, degranulation and thrombosis through complex formation with VEGF and activation of FcγRIIa receptor. This mechanism may be relevant to the thromboembolic complications observed in patients receiving Bev therapy.

2003 ◽  
Vol 77 (4) ◽  
pp. 2768-2774 ◽  
Author(s):  
Bernd Hauck ◽  
Weidong Xiao

ABSTRACT Muscle is an attractive target for gene delivery because of its mass and because vectors can be delivered in a noninvasive fashion. Adeno-associated virus (AAV) has been shown to be effective for muscle-targeted gene transfer. Recent progress in characterization of AAV serotype 1 (AAV1) and AAV6 demonstrated that these two AAV serotypes are far more efficient in transducing muscle than is the traditionally used AAV2. Since all cis elements are identical in these vectors, the potential determinants for their differences in transducing muscle appear to be located within the AAV capsid proteins. In the present study, a series of AAV capsid mutants were generated to identify the major regions affecting AAV transduction efficiency in muscle. Replacement of amino acids 350 to 736 of AAV2 VP1 with the corresponding amino acids from VP1 of AAV1 resulted in a hybrid vector that behaved very similarly to AAV1 in vitro and in vivo in muscle. Characterization of additional mutants carrying smaller regions of the AAV1 VP1 amino acid sequence in the AAV2 capsid protein suggested that amino acids 350 to 430 of VP1 function as a major tissue tropism determinant. Further analysis showed that the heparin binding domain and the major antigenic determinants in the AAV capsid region were not necessary for the efficiency of AAV1 transduction of muscle.


Blood ◽  
1992 ◽  
Vol 79 (8) ◽  
pp. 1995-2003 ◽  
Author(s):  
C Legrand ◽  
V Thibert ◽  
V Dubernard ◽  
B Begault ◽  
J Lawler

Abstract We have investigated the molecular requirements for thrombospondin (TSP) to bind to the platelet surface and to support the subsequent secretion-dependent platelet aggregation. For this, we used two distinct murine monoclonal antibodies (MoAbs), designated MAI and MAII, raised against human platelet TSP, and three polyclonal antibodies, designated R3, R6, and R5, directed against fusion proteins containing the type 1 (Gly 385-Ile 522), type 2 (Pro 559-Ile 669), and type 3 (Asp 784-Val 932) repeating sequences, respectively. Among them, R5 and R6, but not R3, inhibited thrombin-induced aggregation of washed platelets and the concomitant secretion of serotonin. These antibodies, however, did not inhibit the expression of TSP on thrombin-activated platelets, as measured by the binding of a radiolabeled MoAb to TSP, suggesting that they may inhibit platelet aggregation by interfering with a physiologic event subsequent to TSP binding. In contrast, MoAb MAII, which reacts with an epitope located within the heparin-binding domain of TSP, inhibited both TSP surface expression and platelet aggregation/secretion induced by thrombin. In addition, this MoAb inhibited in a dose-dependent manner (IC50 approximately 0.5 mumol/L) the interaction of 125I-TSP with immobilized fibrinogen and platelet glycoprotein IV, both potential physiologic receptors for TSP on thrombin-activated platelets. These results indicate that the interaction of TSP with the surface of activated platelets can be modulated at the level of a specific epitope located within the amino terminal heparin-binding domain of the molecule. Thus, selective inhibition of the platelet/TSP interaction may represent an alternative approach to the inhibition of platelet aggregation.


Blood ◽  
1992 ◽  
Vol 79 (8) ◽  
pp. 1995-2003
Author(s):  
C Legrand ◽  
V Thibert ◽  
V Dubernard ◽  
B Begault ◽  
J Lawler

We have investigated the molecular requirements for thrombospondin (TSP) to bind to the platelet surface and to support the subsequent secretion-dependent platelet aggregation. For this, we used two distinct murine monoclonal antibodies (MoAbs), designated MAI and MAII, raised against human platelet TSP, and three polyclonal antibodies, designated R3, R6, and R5, directed against fusion proteins containing the type 1 (Gly 385-Ile 522), type 2 (Pro 559-Ile 669), and type 3 (Asp 784-Val 932) repeating sequences, respectively. Among them, R5 and R6, but not R3, inhibited thrombin-induced aggregation of washed platelets and the concomitant secretion of serotonin. These antibodies, however, did not inhibit the expression of TSP on thrombin-activated platelets, as measured by the binding of a radiolabeled MoAb to TSP, suggesting that they may inhibit platelet aggregation by interfering with a physiologic event subsequent to TSP binding. In contrast, MoAb MAII, which reacts with an epitope located within the heparin-binding domain of TSP, inhibited both TSP surface expression and platelet aggregation/secretion induced by thrombin. In addition, this MoAb inhibited in a dose-dependent manner (IC50 approximately 0.5 mumol/L) the interaction of 125I-TSP with immobilized fibrinogen and platelet glycoprotein IV, both potential physiologic receptors for TSP on thrombin-activated platelets. These results indicate that the interaction of TSP with the surface of activated platelets can be modulated at the level of a specific epitope located within the amino terminal heparin-binding domain of the molecule. Thus, selective inhibition of the platelet/TSP interaction may represent an alternative approach to the inhibition of platelet aggregation.


Structure ◽  
1998 ◽  
Vol 6 (5) ◽  
pp. 637-648 ◽  
Author(s):  
Wayne J Fairbrother ◽  
Mark A Champe ◽  
Hans W Christinger ◽  
Bruce A Keyt ◽  
Melissa A Starovasnik

Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 2611-2611
Author(s):  
Amr El-Sheikh ◽  
Gourab Bhattacharjee ◽  
Per Borgstrom ◽  
Mattias Belting ◽  
Thomas Edgington

Abstract The expression of certain proteoglycans or modification of proteins within the vascular tree, dependent on the microenvironment, is critical for elucidating the biology of endothelium specificity and development. It also facilitates the targeting of physiologic and therapeutic agents to different addresses within the vascular map. Using in vivo panning, we have identified a truncated heparin-binding domain (HBDt) that recognizes its target selectively in tumor vasculature. Here we show that it localizes selectively to the endothelial cells of intra-tumoral blood vessels of various murine tumor models, such as CT26, LLC, N202, and Tramp-L1. The HBDt, as a part of the VEGF heparin-binding domain, is conserved throughout evolution and is known to bind the VEGFR-2/Npn-1 complex. Although the VEGFR-2/Npn-1 complex is expressed elsewhere in the vascular tree, this domain only localizes to a target in tumor vasculature. We have analyzed the basis of this selectivity in vitro and in vivo. In vitro analysis has shown that chondroitin sulfates are the most potent inhibitors of HBDt binding to heparin. We also show, using Western blot and confocal microscopy analyses, that VEGFR-2 and Npn-1, although expressed in different organs, are only recognized by HBDt when coexpressed with chondroitin sulfate C (C6S) in the tumor vasculature. The HBDt colocalized with VEGFR-2, Npn-1, and C6S but with not bFGFR or heparan sulfates in the intravasculature of different tumor models. Furthermore, the selective expression of C6S oligosaccaharide, in conjunction with VEGFR-2 and Npn-1, during the angiogenesis of tumor endothelium defines the target for the HBDt but not during aortic angiogensis. Therefore, our data demonstrate that the expression of C6S, as part of the HBDt receptor, is an example of the tumor microenvironment conditioning, which imparts association of a novel target on endothelium surfaces of tumors.


1994 ◽  
Vol 300 (2) ◽  
pp. 443-448 ◽  
Author(s):  
E Vilella ◽  
G Bengtsson-Olivecrona ◽  
T Stigbrand ◽  
P E Jensen

The interaction between bovine lipoprotein lipase (bLPL) and human alpha 2-macroglobulin (alpha 2M) was studied by use of non-denaturing PAGE and gel-permeation, Zn(2+)-Sepharose and heparin-Sepharose chromatography. It was demonstrated that bLPL in vitro binds non-covalently to native alpha 2M, but not to the receptor-recognized form produced by treatment of alpha 2M with chymotrypsin or methylamine. A small amount of bLPL was bound covalently to alpha 2M by disulphide interchange, when incubated together with chymotrypsin or methylamine. Whereas alpha 2M in complex with bLPL still bound to Zn(2+)-Sepharose, bLPL lost the ability to bind to heparin-Sepharose. Preincubation of bLPL with heparin prevented complex-formation with alpha 2M, suggesting that alpha 2M interacts with the heparin-binding domain of bLPL. Experiments in which 125I-bLPL was incubated with human plasma at 20 degrees C demonstrated an 11-17% binding of the labelled lipase to alpha 2M, indicating that this interaction may be of physiological significance.


Sign in / Sign up

Export Citation Format

Share Document